Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Heliyon ; 7(8): e07749, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34430738

RESUMEN

RAPTA-EA1 is a promising glutathione transferase (GSTP-1) inhibitor that has previously been shown to inhibit the growth of various breast cancer cells. We studied the anticancer activity of RAPTA-EA1 on triple-negative BRCA1 competent breast cancer MDA-MB-231 cells. MDA-MB-231 cells are significantly more sensitive to RAPTA-EA1 than MCF-7 cells. Treatment reveals a higher degree of cytotoxicity than cisplatin against both cell lines. Ruthenium accumulation in MDA-MB-231 cells is mainly in the nuclear fraction (43%), followed by the cytoplasm (30%), and the mitochondria (27%). RAPTA-EA1 blocks cell growth at the G2/M phase, leading to nuclear condensation and cell death. The compound slightly inhibits DNA replication of the 3,426-bp fragment of the BRCA1 exon 11 of the cells, with approximately 0.6 lesion per the BRCA1 fragment. The expression of BRCA1 mRNA and its protein in the Ru-treated cells is curtailed by 50-80% compared to the untreated controls. Growth inhibition of the triple-negative BRCA1 wild-type MDA-MB-231 and the sporadic BRCA1 wild-type MCF-7 cells by olaparib (a poly [ADP-ribose] polymerase (PARP) inhibitor) is dose-dependent, with MDA-MB-231 cells being two-fold less susceptible to the drug than MCF-7 cells. Combining olaparib with RAPTA-EA1 results in a combination index (CI) of 0.78 (almost additive) in MDA-MB-231 cells and 0.24 (potent synergy) in the MCF-7 cells. The PARP inhibitor alone differently regulates the expression of BRCA1 mRNA in both cell lines, whereas the olaparib-RAPTA-EA1 combination induces overexpression of BRCA1 mRNA in these cells. However, the expression level of the BRCA1 protein is dramatically reduced after treatment with the combined inhibitors, compared with the untreated controls. This observation highlights the cellular responses of triple-negative BRCA1 proficient breast cancer MDA-MB-231 cells to RAPTA-EA1 through BRCA1 inhibition and provides insights into alternative treatments for breast cancer.

2.
Apoptosis ; 24(7-8): 612-622, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31016421

RESUMEN

An organometallic ruthenium(II) arene compound, Ru(η6-toluene)(PTA)Cl2 (PTA = 1,3,5-triaza-7-phosphaadamantane), termed RAPTA-T, exerts promising antimetastatic properties. In this study, the effects of RAPTA-T on BRCA1-defective HCC1937 breast cancer cells have been investigated, and compared to its effects on BRCA1-competent MCF-7 breast cancer cells. RAPTA-T showed a very low cytotoxicity against both tested cells. Ruthenium is found mostly in the cytoplasmic compartment of both cells. Flow cytometric analysis reveals that the compound arrests the growth of both cells by triggering the G2/M phase that led to the induction of apoptosis. At equimolar concentrations, RAPTA-T causes much more cellular BRCA1 damage in HCC1937 than in MCF-7 cells, suppressing the expression of BRCA1 mRNA in both cell lines with the subsequent down-regulation of the BRCA1 protein. Interestingly, RAPTA-T exhibits an approximately fivefold greater ability to suppress the expression of the BRCA1 protein in HCC1937 than in MCF-7 cells. These data provide insights into the molecular mechanisms by which RAPTA-T exerts its effects on BRCA1-associated breast cancer cells.


Asunto(s)
Antineoplásicos/farmacología , Proteína BRCA1/genética , Neoplasias de la Mama/genética , Compuestos Organometálicos/farmacología , Antineoplásicos/química , Antineoplásicos/metabolismo , Apoptosis/efectos de los fármacos , Proteína BRCA1/metabolismo , Neoplasias de la Mama/patología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Citoplasma/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Células MCF-7 , Compuestos Organometálicos/química , Compuestos Organometálicos/metabolismo , Rutenio/química , Rutenio/metabolismo
3.
Biochem Biophys Res Commun ; 488(2): 355-361, 2017 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-28501617

RESUMEN

RAPTA compounds, ([Ru(η6-arene)(PTA)Cl2], PTA = 1,3,5-triaza-7-phosphaadamantane), have been reported to overcome drug resistance in cisplatin resistant cells. However, the exact mechanism of these complexes is still largely unexplored. In this study, the interaction of some RAPTA compounds with the N-terminal fragment of the BRCA1 RING domain protein was investigated. The binding of the RAPTA compounds to the BRCA1 protein resulted in a release of Zn2+ ions in a dose and time dependent manner, as well as thermal alteration of ruthenated-BRCA1 proteins. Electron Transfer Dissociation (ETD) fragmentation mass spectrometry revealed the preferential binding sites of the RAPTA complexes on the BRCA1 zinc finger RING domain at a similar short peptide stretch, Cys24Lys25Phe26Cys27Met28Leu29 and Lys35 (residues 44-49 and 55 on full length BRCA1). Changes in the conformation and binding constants of ruthenium-BRCA1 adducts were established, resulting in inactivation of the RING heterodimer BRCA1/BARD1-mediated E3 ubiquitin ligase function. These findings could provide mechanistic insight into the mode of action of RAPTA complexes for on tested BRCA1 model protein.


Asunto(s)
Adamantano/análogos & derivados , Proteína BRCA1/metabolismo , Compuestos Organofosforados/farmacología , Dominios RING Finger/efectos de los fármacos , Ubiquitina-Proteína Ligasas/metabolismo , Adamantano/química , Adamantano/farmacología , Proteína BRCA1/antagonistas & inhibidores , Proteína BRCA1/química , Sitios de Unión/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Estructura Molecular , Compuestos Organofosforados/química , Relación Estructura-Actividad , Ubiquitina-Proteína Ligasas/antagonistas & inhibidores
4.
Anticancer Agents Med Chem ; 17(2): 212-220, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27039925

RESUMEN

BACKGROUND: The RAPTA-EA1 complex [ruthenium(II)-arene 1,3,5-triaza-7-phosphaadamantane (pta) complex with an arene-tethered ethacrynic acid ligand] has been reported to overcome drug resistance that developed due to the current use of platinum-based treatments. However, the exact mechanism of action of RAPTA-EA1 remains largely unexplored and unknown. OBJECTIVE: Here we have further studied the effect of RAPTA-EA1 on BRCA1-defective HCC1937 breast cancer cells and compared its effects on BRCA1-competent MCF-7 breast cancer cells. METHOD: HCC1937 and MCF-7 breast cancer cells were treated with the RAPTA-EA1 complex. The cytotoxicity of ruthenium-induced cells was evaluated by a MTT assay. Cellular uptake of ruthenium was determined by ICP-MS. Cell cycle and apoptosis were assessed using a flow cytometer. Expression of BRCA1 mRNA and its encoded protein was quantitated by a real-time RT-PCR and Western blotting. RESULTS: Differences in cytotoxicity were correlated with the differential accumulations of ruthenium and the induction of apoptosis. The ruthenium complex caused dramatically more damage to the BRCA1 gene in the BRCA1-defective HCC1937 cells than to the BRCA1-competent MCF-7 cells. It decreased the expression of BRCA1 mRNA in the BRCA1-competent cells, while in contrast, its expression increased in the BRCA1-defective cells. However, the expression of the BRCA1 protein was significantly reduced in both types of breast cancer cells. CONCLUSION: The results presented here have demonstrated a differential cellular response for the BRCA1-defective and BRCA1-competent breast cancer cells to RAPTA-EA1. These findings have provided more insight into the actions and development of the ruthenium-based compounds for use for the treatment of breast cancer.


Asunto(s)
Antineoplásicos/farmacología , Proteína BRCA1/genética , Neoplasias de la Mama/tratamiento farmacológico , Regulación hacia Abajo/efectos de los fármacos , Compuestos Organometálicos/farmacología , Rutenio/farmacología , Antineoplásicos/química , Antineoplásicos/farmacocinética , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Femenino , Genes BRCA1/efectos de los fármacos , Humanos , Células MCF-7 , Compuestos Organometálicos/química , Compuestos Organometálicos/farmacocinética , ARN Mensajero/genética , Rutenio/química , Rutenio/farmacocinética
5.
Curr Cancer Drug Targets ; 16(7): 606-17, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26845433

RESUMEN

Triple-negative breast cancer (TNBC) is defined by the absence of expression of estrogen receptor (ER), progesterone receptor (PR), and a lack of overexpression or amplification of human epidermal growth factor receptor 2 (HER2). The clinicopathological characteristics of TNBC include a high grading, a high rate of cell proliferation and a greater degree of chromosomal rearrangement. Patients with triple-negative breast cancer are more likely to be drug resistant and more difficult to treat, and are also frequently BRCA1 mutants. Methylation of the BRCA1 promoter region is associated with a reduction of the BRCA1 mRNA level. TNBC patients with a methylated BRCA1 had a better disease-free survival compared with those with non-methylated BRCA1. From a therapeutic perspective, the expression level of BRCA1 has been a major determinant of the responses to different classes of chemotherapy. BRCA1-dysfunctional tumors are hypersensitive to DNA damaging chemotherapeutic agents like platinum drugs. Although platinum based drugs are currently widely used as conventional chemotherapeutic drugs in breast cancer chemotherapy, their use has several disadvantages. It is therefore of interest to seek out alternative therapeutic metal-based compounds that could overcome the limitations of these platinum based drugs. Ruthenium-based compounds could be the most promising alternative to the platinum drugs. This review highlights the use of BRCA1 as a predictive marker as well as for a potential drug target for anticancer ruthenium compounds.


Asunto(s)
Antineoplásicos/uso terapéutico , Proteína BRCA1/metabolismo , Rutenio/uso terapéutico , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/metabolismo , Femenino , Humanos , Receptor ErbB-2/metabolismo , Receptores de Estrógenos/metabolismo , Receptores de Progesterona/metabolismo
6.
Breast Cancer (Auckl) ; 8: 51-6, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24678242

RESUMEN

The breast cancer susceptibility gene 1 (BRCA1) has been shown to maintain genomic stability through multiple functions in the regulation of DNA damage repair and transcription. Its translated BRCT (BRCA1 C-terminal domain) acts as a strong transcriptional activator. BRCA1 damaged by carboplatin treatment may lead to a loss of such functions. To address the possibility of the BRCA1 gene as a therapeutic target for carboplatin, we investigated the functional consequences of the 3'-terminal region of human BRCA1 following in vitro platination with carboplatin. A reduction in cellular BRCA1 repair of carboplatin-treated plasmid DNA, using a host cell reactivation assay, was dependent on the platination levels on the reporter gene. The transcriptional transactivation activity of the drug-modified BRCA1, assessed using a one-hybrid GAL4 transcriptional assay, was inversely proportional to the carboplatin doses. The data emphasized the potential of the BRCA1 gene to be a target for carboplatin treatment.

7.
BMC Cancer ; 14: 73, 2014 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-24507701

RESUMEN

BACKGROUND: Triple-negative breast cancer (TNBC) is defined by the absence of expression of estrogen receptor, progesterone receptor and human epidermal growth factor receptor 2. Breast cancers with a BRCA1 mutation are also frequently triple-negative. Currently, there is a lack of effective therapies and known specific molecular targets for this aggressive breast cancer subtype. To address this concern, we have explored the cellular responses of BRCA1-defective and triple-negative breast cancer cells, and in vitro BRCA1 interactions induced by the ruthenium(II) complexes containing the bidentate ligand, 5-chloro-2-(phenylazo)pyridine. METHODS: Triple-negative MDA-MB-231, BRCA1-defective HCC1937 and BRCA1-competent MCF-7 breast cancer cell lines were treated with ruthenium(II) complexes. The cytoxoxicity of ruthenium-induced breast cancer cells was evaluated by a real time cellular analyzer (RTCA). Cellular uptake of ruthenium complexes was determined by ICP-MS. Cell cycle progression and apoptosis were assessed using propidium iodide and Annexin V flow cytometry. The N-terminal BRCA1 RING protein was used for conformational and functional studies using circular dichroism and in vitro ubiquitination. RESULTS: HCC1937 cells were significantly more sensitive to the ruthenium complexes than the MDA-MB-231 and MCF-7 cells. Treatment demonstrated a higher degree of cytotoxicity than cisplatin against all three cell lines. Most ruthenium atoms were retained in the nuclear compartment, particularly in HCC1937 cells, after 24 h of incubation, and produced a significant block at the G2/M phase. An increased induction of apoptotic cells as well as an upregulation of p53 mRNA was observed in all tested breast cancer cells. It was of interest that BRCA1 mRNA and replication of BRCA1-defective cells were downregulated. Changes in the conformation and binding constants of ruthenium-BRCA1 adducts were observed, causing inactivation of the RING heterodimer BRCA1/BARD1-mediated E3 ubiquitin ligase activity. CONCLUSIONS: This study has revealed the ability of ruthenium complexes to inhibit cell proliferation, induce cell cycle progression and apoptosis. Ruthenium treatment upregulated the marker genes involved in apoptosis and cell cycle progression while it downregulated BRCA1 mRNA and replication of HCC1937 cells. Our results could provide an alternative approach to finding effective therapeutic ruthenium-based agents with promising anticancer activity, and demonstrated that the BRCA1 RING domain protein was a promising therapeutic target for breast cancers.


Asunto(s)
Antineoplásicos/farmacología , Sustancias Intercalantes/farmacología , Piridinas/farmacología , Rutenio/farmacología , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología , Ubiquitina-Proteína Ligasas/metabolismo , Antineoplásicos/metabolismo , Antineoplásicos/uso terapéutico , Ciclo Celular/efectos de los fármacos , Ciclo Celular/fisiología , Femenino , Humanos , Sustancias Intercalantes/uso terapéutico , Células MCF-7 , Piridinas/química , Piridinas/uso terapéutico , Rutenio/química , Rutenio/uso terapéutico , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico
8.
Int J Mol Sci ; 13(11): 14898-916, 2012 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-23203101

RESUMEN

BRCA1 is a key mediator of DNA repair pathways and participates in the maintenance of the genomic integrity of cells. The control of DNA damage repair mechanisms by BRCA1 is of great interest since molecular defects in this pathway may reflect a predictive value in terms of a cell's sensitivity to DNA damaging agents or anticancer drugs. BRCA1 has been found to exhibit a hormone-dependent pattern of expression in breast cells. Wild-type BRCA1 is required for the inhibition of the growth of breast tumor cells in response to the pure steroidal ERα antagonist fulvestrant. Also a loss of BRCA1-mediated transcriptional activation of ERα expression results in increased resistance to ERα antagonists. Platinum-based drugs, poly(ADP-ribose) polymerase (PARP) inhibitors, and their combination are currently included in chemotherapy regimens for breast cancer. Preclinical and clinical studies in a BRCA1-defective setting have recently indicated a rationale for the use of these compounds against hereditary breast cancers. Initial findings indicate that neoadjuvant use of cisplatin results in high rates of complete pathological response in patients with breast cancer who have BRCA1 mutations. Cisplatin produces a better response in triple-negative breast cancer (TNBC) than in non-TNBC diseases in both the neoadjuvant and adjuvant settings. This implies that TNBC cells may harbor a dysfunctional BRCA1 repair pathway.


Asunto(s)
Proteína BRCA1/genética , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Reparación del ADN , Terapia Molecular Dirigida , Receptores de Estrógenos/genética , Animales , Antineoplásicos Hormonales/farmacología , Antineoplásicos Hormonales/uso terapéutico , Proteína BRCA1/metabolismo , Neoplasias de la Mama/metabolismo , Estrógenos/metabolismo , Femenino , Humanos , Unión Proteica , Receptores de Estrógenos/metabolismo , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/metabolismo
9.
Int J Mol Sci ; 13(10): 13183-202, 2012 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-23202946

RESUMEN

The ruthenium-based complex [Ru(η(6)-p-phenylethacrynate)Cl(2)(pta)] (pta = 1,3,5-triaza-7-phosphatricyclo-[3.3.1.1]decane), termed ethaRAPTA, is an interesting antitumor compound. The elucidation of the molecular mechanism of drug activity is central to the drug development program. To this end, we have characterized the ethaRAPTA interaction with DNA, including probing the sequence specific modified DNA structural stability and DNA amplification using the breast cancer suppressor gene 1 (BRCA1) of human breast and colon adenocarcinoma cell lines as models. The preference of ethaRAPTA base binding is in the order A > G > T > C. Once modified, the ethaRAPTA-induced BRCA1 structure has higher thermal stability than the modified equivalents of its related compound, RAPTA-C. EthaRAPTA exhibits a higher efficiency than RAPTA-C in inhibiting BRCA1 amplification. With respect to both compounds, the inhibition of BRCA1 amplification is more effective in an isolated system than in cell lines. These data provide evidence that will help to understand the process of elucidating the pathways involved in the response induced by ethaRAPTA.


Asunto(s)
Antineoplásicos/farmacología , Daño del ADN/efectos de los fármacos , Compuestos Organometálicos/farmacología , Antineoplásicos/química , Proteína BRCA1/química , Proteína BRCA1/metabolismo , Secuencia de Bases , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Cimenos , ADN/química , ADN/metabolismo , Aductos de ADN/química , Aductos de ADN/metabolismo , Femenino , Células HT29 , Humanos , Células MCF-7 , Datos de Secuencia Molecular , Compuestos Organometálicos/química , Unión Proteica/efectos de los fármacos , Temperatura
10.
Breast Cancer (Auckl) ; 5: 201-8, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22084573

RESUMEN

BRCA1 is a tumor suppressor protein involved in maintaining genomic integrity through multiple functions in DNA damage repair, transcriptional regulation, cell cycle checkpoint, and protein ubiquitination. The BRCA1-BARD1 RING complex has an E3 ubiquitin ligase function that plays essential roles in response to DNA damage repair. BRCA1-associated cancers have been shown to confer a hypersensitivity to chemotherapeutic agents. Here, we have studied the functional consequence of the in vitro E3 ubiquitin ligase activity and cisplatin sensitivity of the missense mutation D67Y BRCA1 RING domain. The D67Y BRCA1 RING domain protein exhibited the reduced ubiquitination function, and was more susceptible to the drug than the D67E or wild-type BRCA1 RING domain protein. This evidence emphasized the potential of using the BRCA1 dysfunction as an important determinant of chemotherapy responses in breast cancer.

12.
Breast Cancer Res Treat ; 126(1): 203-9, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20878461

RESUMEN

The breast cancer susceptibility protein 1 (BRCA1) participates in the maintenance of cells genomic integrity through DNA repair, cell cycle checkpoint, protein ubiquitination, and transcriptional regulation. The N-terminus of BRCA1 contains a RING domain that preferentially forms a heterodimeric complex with BARD1. The BRCA1-BARD1 RING complex has an E3 ubiquitin ligase activity that plays an essential role in response to DNA damage. Preclinical and clinical studies have recently revealed that structural changes to the heterodimer result in alterations to the BRCA1-mediated DNA repair pathways in cancer cells, and lead to hypersensitivity to several chemotherapeutic agents. It is of interest to approach the BRCA1 RING domain as a potentially molecular target for platinum-based drugs for cancer therapy. A previous study has shown that the anticancer drug cisplatin formed intramolecular and intermolecular BRCA1 adducts in which His117 was the primary platinum-binding site, and conferred conformational changes and induced thermostability. Here, we have studied the functional consequence of the in vitro platination of the BRCA1 RING domain by a number of platinum complexes. The BRCA1 ubiquitin ligase activity was inhibited by transplatin > cisplatin > oxaliplatin > carboplatin in that order. The consequences of the binding of the platinum complexes on the reactivity of the BRCA1 were also discussed. The data raised the possibility of selectively targeting the BRCA1 DNA repair for cancer therapy.


Asunto(s)
Proteína BRCA1/antagonistas & inhibidores , Neoplasias de la Mama/metabolismo , Carboplatino/farmacología , Cisplatino/farmacología , Compuestos Organoplatinos/farmacología , Proteínas Supresoras de Tumor/antagonistas & inhibidores , Ubiquitina-Proteína Ligasas/antagonistas & inhibidores , Antineoplásicos/farmacología , Proteína BRCA1/metabolismo , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Reactivos de Enlaces Cruzados/farmacología , Femenino , Humanos , Oxaliplatino , Conformación Proteica , Multimerización de Proteína , Estructura Terciaria de Proteína , Células Tumorales Cultivadas , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
13.
J Biol Inorg Chem ; 16(2): 217-26, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20967475

RESUMEN

Breast cancer susceptibility protein 1 (BRCA1) participates in genomic integrity maintenance through DNA repair, cell cycle checkpoint, protein ubiquitination, and transcriptional regulation. The N-terminus of BRCA1 contains a RING domain which forms two Zn(2+) binding sites in an interleaved fashion. A number of deleterious BRCA1 missense mutations, which predispose an individual to a subset of hereditary breast and ovarian cancers, have been identified in the RING domain. Disruption of Zn(2+) binding sites and protein structure results in the inactivation of BRCA1 tumor suppression function. An unprecedented D67E BRCA1 mutation, identified in Thai familial breast cancer patients, is located in the vicinity of Zn(2+) binding site II, and its pathogenic significance remains elusive. The present study revealed that the D67E BRCA1 RING protein assumes a preformed structure in the absence of Zn(2+). The Zn(2+)-bound mutant protein was more folded, resulting in enhanced proteolytic resistance and dimerization. This indicated that the mutation retained Zn(2+) binding, and barely perturbed the native global structure of the BRCA1 RING domain. The complex between D67E BRCA1 and BARD1 RING domains exhibited a substantial ubiquitin ligase activity compared with a defective complex containing the C61G BRCA1 mutation. However, the D67E mutation was slightly less stable toward thermal denaturation. This implies that the D67E mutation might be a neutral or mild cancer-risk modifier of other defective mechanisms underlying BRCA1-mutation-related breast cancer.


Asunto(s)
Ácido Aspártico/química , Proteína BRCA1/química , Proteína BRCA1/metabolismo , Ácido Glutámico/química , Ácido Aspártico/genética , Proteína BRCA1/genética , Cromatografía en Gel , Dicroismo Circular , Femenino , Ácido Glutámico/genética , Humanos , Espectrometría de Masas , Unión Proteica/genética , Unión Proteica/fisiología , Relación Estructura-Actividad , Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Espectroscopía de Absorción de Rayos X , Zinc/metabolismo
14.
Chem Biodivers ; 7(8): 1949-67, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20730959

RESUMEN

The breast cancer suppressor protein 1 (BRCA1) has been shown to participate in genomic integrity maintenance. Preclinical and clinical studies have recently revealed that the inactivation of BRCA1 in cancer cells leads to chemosensitivity. Approaching the BRCA1 RING protein as a potentially molecular target for a platinum-based drug might be of interest in cancer therapy. In the present study, the in vitro platination of the BRCA1 RING protein by the anticancer drug cisplatin was observed. The protein contained a preformed structure in the apo form with structural changes and resistance to limited proteolysis after Zn2+ binding. SDS-PAGE and mass-spectrometric analyses revealed that cisplatin preferentially formed monofunctional and bifunctional BRCA1 adducts. Tandem mass spectrometry (MS/MS) of the 656.29(2+) ion indicated that the ion arose from [Pt(NH3)2(OH)]+ bound to the BRCA1 peptide (111)ENNSPEHLK(119). The product-ion spectrum revealed the Pt-binding site on His117. Circular dichroism showed that the apo form, not holo form, of BRCA1 underwent more folded structural rearrangement upon cisplatin binding. Cisplatin-bound protein exhibited an enhanced thermostability by 13 degrees , resulting from the favorably intermolecular cross-links driven by the free energy. Our findings demonstrated the first conformational and thermal evidences for a direct binding of cisplatin to the BRCA1 RING domain and could raise a possibility of selectively targeted treatment of cancer with less toxicity or improved response to conventional regimens.


Asunto(s)
Proteína BRCA1/química , Cisplatino/farmacología , Dominios RING Finger/efectos de los fármacos , Secuencia de Aminoácidos , Antineoplásicos/farmacología , Proteína BRCA1/metabolismo , Dicroismo Circular , Cisplatino/metabolismo , Dimerización , Estabilidad de Medicamentos , Humanos , Datos de Secuencia Molecular , Temperatura
15.
Chem Biodivers ; 7(5): 1290-302, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20491084

RESUMEN

The interaction of two ruthenium-arene-1,3,5-triaza-7-phosphaadamantane compounds ([Ru(eta(6)-p-cymene)Cl(2)(pta)] and [Ru(eta(6)-p-cymene)(C(6)H(6)O(4))(pta)], termed RAPTA-C (3) and carboRAPTA-C (4), resp.) with the DNA sequence of the human breast-cancer suppressor gene 1 (BRCA1) has been studied using a range of techniques that probe conformation, cross-linking, base specificity, restriction analysis, and in vitro inhibition of DNA polymerization. The study demonstrates that substitution of the two labile chloride ligands in 3 by the more stable cyclobutane-1,1-dicarboxylate ligand onto the RAPTA framework reduces the rate of reaction with DNA in a similar manner to the analogous Pt-based drug pair cisplatin (1) and carboplatin (2), suggesting that hydrolysis may be a prerequisite to DNA binding with the Ru compounds. Moreover, the rate of DNA interaction for 3 is in a similar range to that of 2, despite the fact that these compounds have a different therapeutic profile. The similar rates of reaction contrasting with the different modes of activity suggests that the RAPTA compounds may be clinically useful against cancer cells that have developed resistance to Pt-based therapies, particularly involving excision-repair mechanisms.


Asunto(s)
Antineoplásicos/farmacología , Proteína BRCA1/genética , Complejos de Coordinación/farmacología , Compuestos Organometálicos/farmacología , Antineoplásicos/química , Proteína BRCA1/química , Secuencia de Bases , Complejos de Coordinación/química , Cimenos , ADN/química , Aductos de ADN/análisis , Aductos de ADN/química , Daño del ADN , Humanos , Datos de Secuencia Molecular , Compuestos Organometálicos/química
16.
Cancer Biol Ther ; 8(10): 890-8, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19270496

RESUMEN

BRCA1 is a tumor suppressor gene. Its translated product has an important function in transcriptional activation and DNA repair pathways. Damaged BRCA1 due to cisplatin treatment may lead to loss of such functions. To address a potential drug target of BRCA1 for cisplatin treatment, we investigated the biophysical characterization and functional consequences of the 3'-terminal region of human BRCA1 after in vitro platination with cisplatin. To analyze the base/sequence specificity of cisplatin damage, the measurement for sensitivity of cisplatin-treated BRCA1 to restriction enzymes (EcoO109I and PvuII) and sequence gel analysis was conducted. The results suggested that the platination favorably occurred at the d(GpG) and the d(GpC) sites. An increase in drug concentrations resulted in increased interstrand crosslinks at the d(GpC) site. Cisplatin affected the transition temperature of the BRCA1 gene fragment in a biphasic fashion. DSC thermogram of DNA adducts was shifted to a lower transition temperature at lower cisplatin concentration. However, at higher drug concentration, the thermogram peaked at a slightly higher transition temperature with predominantly increased heat specific capacity. Reduction in cellular DNA repair of cisplatin-damaged plasmid DNA, using host cell reactivation assay, was a consequence of an increase in platination levels on the reporter gene. The GAL4-fused BRCA1 slightly enhanced the transcription of the reporter gene in the absence of GAL4 binding site. The transcriptional transactivation activity of cisplatin-modified BRCA1, when tested in "one-hybrid GAL4 transcriptional assay," was inversely proportional to cisplatin doses. Furthermore, the transcriptional transactivation activity was dramatically diminished in the presence of a second expression vector containing multiple cisplatin-damaged sites. The data provide the first evidence for direct interaction of cisplatin with BRCA1 and raise the possibility of BRCA1 as a therapeutic target for platinum drug-based chemotherapy.


Asunto(s)
Antineoplásicos/metabolismo , Cisplatino/metabolismo , Daño del ADN/efectos de los fármacos , Genes BRCA1/fisiología , Activación Transcripcional , Neoplasias de la Mama/metabolismo , Rastreo Diferencial de Calorimetría , Línea Celular Tumoral , Reparación del ADN , Relación Dosis-Respuesta a Droga , Femenino , Genes Reporteros , Humanos , Luciferasas de Renilla/metabolismo , Plásmidos/genética , Temperatura , Transfección
17.
Biochim Biophys Acta ; 1725(2): 145-51, 2005 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-16099593

RESUMEN

Carboplatin is an anticancer drug for the treatment of cancers affecting various organs including ovary and testes. It essentially exerts its cytotoxicity against cancerous cells via covalent attachment of platinum atom to DNA, generating various platinum-DNA adducts. Platinum-DNA adducts inhibit biological processes essential for cellular viability. However, carboplatin interacts nonspecifically with DNA, resulting in damaging of normal cell DNA. Potential in vitro interaction of carboplatin with genes encoding tumor suppressor proteins such as human breast cancer suppressor gene 1(BRCA1) was herein investigated. The 696--bp fragment of the 3'-region of BRCA1 gene (nucleotide 4897--5592) was amplified by RT-PCR using mRNA templates isolated from human white blood cells. Retardation of the electrophoretic migration on agarose gel of drug-treated DNA, in the dose-response manner, was observed. Analysis by restriction digestion with PvuII and Eco O 109I suggested that the platination favorably occurred at the dGpG sequence of Eco O 109I-cleaved site. The semi-quantitative PCR-based assay was used to determine the lesion frequencies produced by carboplatin in the 696-bp fragment of the 3'-region of BRCA1 gene and in the 3,426-bp fragment of the BRCA1 exon 11 of human breast adenocarcinoma MCF-7 cells. A significant decrease in DNA amplification was observed at 400 microM of carboplatin with approximately 1--2 platinum atoms per BRCA1 fragment. Carboplatin caused slightly less damage at equimolar concentrations in cells than in cell-free BRCA1 fragment.


Asunto(s)
Proteína BRCA1/química , Neoplasias de la Mama/química , Carboplatino/administración & dosificación , Carboplatino/química , Daño del ADN/efectos de los fármacos , ADN de Neoplasias/química , Compuestos Organoplatinos/química , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Aductos de ADN/efectos de los fármacos , ADN de Neoplasias/genética , ADN de Neoplasias/metabolismo , Relación Dosis-Respuesta a Droga , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Compuestos Organoplatinos/metabolismo
18.
Int J Oncol ; 22(2): 325-32, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12527930

RESUMEN

Dihydropyrimidine dehydrogenase (DPD) is the first and rate-limiting enzyme in the degradation of pyrimidines and pyrimidine base analogs including the anticancer drugs 5-fluorouracil (5-FU) and Xeloda. A decreased DPD enzyme activity has been described in cancer patients experiencing severe and life-threatening toxicity after 5-FU treatment and distinct sequence variants in the DPD gene (DPYD) have been associated with impaired enzyme function. The most prominent mutation in the DPD deficient patient group, a mutation in the splicing donor consensus sequence of intron 14, IVS14+1g>a, resulting in a truncated protein, has been observed in the Caucasian population at frequencies as high as 0.91%-0.94%. This underlines the need for a test system for DPYD mutations in patients undergoing chemotherapy with 5-FU or with Xeloda. To set up a fast and sensitive method to identify variant DPYD alleles, we analyzed 50 healthy individuals by denaturing high performance liquid chromatography (DHPLC). A primer set spanning the whole coding region and the exon-intron boundaries of DPYD was used. In addition, a cDNA-based assay was developed to rapidly identify the 165 base pair deletion in the corresponding RNA of IVS14+1g>a mutation carriers. The optimal mutation detection was elaborated for each of the PCR fragments. DHPLC analysis detected 5 different genetic alterations occurring in the coding region of the gene, as well as 10 intronic sequence variants, respectively. In conclusion, high-throughput screening for DPYD variants by DHPLC may be a reliable tool in the investigation of the molecular basis of DPD deficiency. Furthermore, it will help to identify patients at risk for toxic side effects upon chemotherapy using 5-FU and will facilitate individual treatment of patients.


Asunto(s)
Antineoplásicos/farmacocinética , Cromatografía Líquida de Alta Presión/métodos , Análisis Mutacional de ADN/métodos , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacocinética , Fluorouracilo/farmacocinética , Genotipo , Oxidorreductasas/genética , Pirimidinas/metabolismo , Adulto , Alelos , Sustitución de Aminoácidos , Antineoplásicos/efectos adversos , Biotransformación , Capecitabina , Dihidrouracilo Deshidrogenasa (NADP) , Exones/genética , Femenino , Fluorouracilo/efectos adversos , Genes , Humanos , Inactivación Metabólica , Intrones/genética , Masculino , Mutación Missense , Oxidorreductasas/deficiencia , Mutación Puntual , Reacción en Cadena de la Polimerasa , Desnaturalización Proteica , Pirimidinas/farmacocinética , Sitios de Empalme de ARN/genética , Sensibilidad y Especificidad , Eliminación de Secuencia , Factores de Tiempo , Población Blanca/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...